Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Immunity ; 54(6): 1200-1218.e9, 2021 06 08.
Article in English | MEDLINE | ID: mdl-33951416

ABSTRACT

Tissue macrophages self-renew during homeostasis and produce inflammatory mediators upon microbial infection. We examined the relationship between proliferative and inflammatory properties of tissue macrophages by defining the impact of the Wnt/ß-catenin pathway, a central regulator of self-renewal, in alveolar macrophages (AMs). Activation of ß-catenin by Wnt ligand inhibited AM proliferation and stemness, but promoted inflammatory activity. In a murine influenza viral pneumonia model, ß-catenin-mediated AM inflammatory activity promoted acute host morbidity; in contrast, AM proliferation enabled repopulation of reparative AMs and tissue recovery following viral clearance. Mechanistically, Wnt treatment promoted ß-catenin-HIF-1α interaction and glycolysis-dependent inflammation while suppressing mitochondrial metabolism and thereby, AM proliferation. Differential HIF-1α activities distinguished proliferative and inflammatory AMs in vivo. This ß-catenin-HIF-1α axis was conserved in human AMs and enhanced HIF-1α expression associated with macrophage inflammation in COVID-19 patients. Thus, inflammatory and reparative activities of lung macrophages are regulated by ß-catenin-HIF-1α signaling, with implications for the treatment of severe respiratory diseases.


Subject(s)
COVID-19/immunology , COVID-19/virology , Cell Self Renewal/immunology , Host-Pathogen Interactions/immunology , Macrophages/immunology , SARS-CoV-2/immunology , Biomarkers , COVID-19/metabolism , Cytokines/metabolism , Disease Susceptibility/immunology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Inflammation Mediators/metabolism , Macrophages/cytology , Macrophages/metabolism , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Signal Transduction
2.
Immunity ; 50(5): 1188-1201.e6, 2019 05 21.
Article in English | MEDLINE | ID: mdl-31053504

ABSTRACT

Lymph nodes (LNs) play critical roles in adaptive immunity by concentrating in one location the antigens, antigen-presenting cells, and antigen-responsive lymphocytes involved in such responses. Recent studies have revealed nonrandom localization of innate and adaptive immune cells within these organs, suggesting that microanatomical positioning optimizes responses involving sparse cooperating cells. Here, we report that the peripheral localization of LN cDC2 dendritic cells specialized for MHC-II antigen presentation is matched by a similarly biased paracortical distribution of CD4+ T cells directed by the chemoattractant receptor Ebi2. In the absence of Ebi2, CD4+ T cells lose their location bias and are delayed in antigen recognition, proliferative expansion, differentiation, direct effector activity, and provision of help for CD8+ T cell-mediated memory responses, limiting host defense and vaccine responses. These findings demonstrate evolutionary selection for distinct niches within the LN that promote cellular responses, emphasizing the critical link between fine-grained tissue organization and host defense.


Subject(s)
Adaptive Immunity/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Receptors, G-Protein-Coupled/metabolism , Animals , Antigen Presentation/immunology , Antigens/immunology , Cell Differentiation/immunology , Histocompatibility Antigens Class II/immunology , Lymph Nodes/cytology , Lymph Nodes/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Receptors, G-Protein-Coupled/genetics
3.
J Immunol ; 202(4): 1033-1038, 2019 02 15.
Article in English | MEDLINE | ID: mdl-30626694

ABSTRACT

Intricate life-versus-death decisions are programmed during T cell development, and the regulatory mechanisms that coordinate their activation and repression are still under investigation. In this study, HDAC3-deficient double-positive (DP) thymocytes exhibit a severe decrease in numbers. The thymic cortex is rich in ATP, which is released by macrophages that clear apoptotic DP thymocytes that fail to undergo positive selection. We demonstrate that HDAC3 is required to repress expression of the purinergic receptor P2X7 to prevent DP cell death. HDAC3-deficient DP thymocytes upregulate the P2X7 receptor, increasing sensitivity to ATP-induced cell death. P2rx7/HDAC3-double knockout mice show a partial rescue in DP cell number. HDAC3 directly binds to the P2rx7 enhancer, which is hyperacetylated in the absence of HDAC3. In addition, RORγt binds to the P2rx7 enhancer and promotes P2X7 receptor expression in the absence of HDAC3. Therefore, HDAC3 is a critical regulator of DP thymocyte survival and is required to suppress P2X7 receptor expression.


Subject(s)
Cell Death , Histone Deacetylases/metabolism , Receptors, Purinergic P2X7/metabolism , Thymocytes/cytology , Thymocytes/enzymology , Animals , Histone Deacetylases/deficiency , Mice , Mice, Knockout , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Receptors, Purinergic P2X7/genetics , Thymocytes/metabolism
4.
Immunity ; 28(3): 425-35, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18328742

ABSTRACT

Costimulatory signals are critical to T cell activation, but how their effects are mediated remains incompletely characterized. Here, we demonstrate that locally produced C5a and C3a anaphylatoxins interacting with their G protein-coupled receptors (GPCRs), C5aR and C3aR, on APCs and T cells both upstream and downstream of CD28 and CD40L signaling are integrally involved in T cell proliferation and differentiation. Disabling these interactions reduced MHC class II and costimulatory-molecule expression and dramatically diminished T cell responses. Importantly, impaired T cell activation by Cd80-/-Cd86-/- and Cd40-/- APCs was reconstituted by added C5a or C3a. C5aR and C3aR mediated their effects via PI-3 kinase-gamma-dependent AKT phosphorylation, providing a link between GPCR signaling, CD28 costimulation, and T cell survival. These local paracrine and autocrine interactions thus operate constitutively in naive T cells to maintain viability, and their amplification by cognate APC partners thus is critical to T cell costimulation.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cell Survival/immunology , Complement C3a/immunology , Complement C5a/immunology , Lymphocyte Activation/immunology , Animals , Antigen Presentation/immunology , Antigen-Presenting Cells/immunology , B7-1 Antigen/immunology , B7-1 Antigen/metabolism , CD28 Antigens/immunology , CD28 Antigens/metabolism , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , CD40 Antigens/immunology , CD40 Antigens/metabolism , CD40 Ligand/immunology , CD40 Ligand/metabolism , Cell Differentiation/immunology , Complement C3a/metabolism , Complement C5a/metabolism , Flow Cytometry , Immunoblotting , Immunoprecipitation , Mice , Mice, Transgenic , Polymerase Chain Reaction , RNA, Messenger/analysis , Receptor, Anaphylatoxin C5a/immunology , Receptor, Anaphylatoxin C5a/metabolism , Signal Transduction/immunology
5.
Science ; 373(6553)2021 07 23.
Article in English | MEDLINE | ID: mdl-34083450

ABSTRACT

The meninges contain adaptive immune cells that provide immunosurveillance of the central nervous system (CNS). These cells are thought to derive from the systemic circulation. Through single-cell analyses, confocal imaging, bone marrow chimeras, and parabiosis experiments, we show that meningeal B cells derive locally from the calvaria, which harbors a bone marrow niche for hematopoiesis. B cells reach the meninges from the calvaria through specialized vascular connections. This calvarial-meningeal path of B cell development may provide the CNS with a constant supply of B cells educated by CNS antigens. Conversely, we show that a subset of antigen-experienced B cells that populate the meninges in aging mice are blood-borne. These results identify a private source for meningeal B cells, which may help maintain immune privilege within the CNS.


Subject(s)
B-Lymphocyte Subsets/physiology , B-Lymphocytes/physiology , Bone Marrow Cells/physiology , Central Nervous System/immunology , Dura Mater/cytology , Lymphopoiesis , Meninges/cytology , Meninges/immunology , Skull/anatomy & histology , Aging , Animals , B-Lymphocyte Subsets/immunology , Cell Movement , Central Nervous System/physiology , Dura Mater/immunology , Fibroblasts/physiology , Homeostasis , Immune Privilege , Mice , Plasma Cells/physiology , Single-Cell Analysis
6.
Mol Immunol ; 45(14): 3756-66, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18614237

ABSTRACT

T cell receptor signaling processes are controlled by the integrated actions of families of protein tyrosine kinases (PTKs) and protein tyrosine phosphatases (PTPases). Several distinct cytosolic protein tyrosine phosphatases have been described that are able to negatively regulate TCR signaling pathways, including SHP-1, SHP-2, PTPH1, and PEP. Using PTPase substrate-trapping mutants and wild type enzymes, we determined that PTPN4/PTP-MEG1, a PTPH1-family member, could complex and dephosphorylate the ITAMs of the TCR zeta subunit. In addition, the substrate-trapping derivative augmented basal and TCR-induced activation of NF-kappaB in T cells. To characterize the contribution of this PTPase in T cells, we developed PTPN4-deficient mice. T cell development and TCR signaling events were comparable between wild type and PTPN4-deficient animals. The magnitude and duration of TCR-regulated ITAM phosphorylation, as well as overall protein phosphorylation, was unaltered in the absence of PTPN4. Finally, Th1- and Th2-derived cytokines and in vivo immune responses to Listeria monocytogenes were equivalent between wild type and PTPN4-deficient mice. These findings suggest that additional PTPases are involved in controlling ITAM phosphorylations.


Subject(s)
NF-kappa B/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 4/physiology , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/physiology , Animals , COS Cells , Cell Line , Chlorocebus aethiops , Humans , Jurkat Cells , Kidney/cytology , Mice , NF-kappa B/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 4/chemistry , Protein Tyrosine Phosphatase, Non-Receptor Type 4/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 4/metabolism , Receptors, Antigen, T-Cell/immunology , Transfection
7.
Mol Immunol ; 45(8): 2380-90, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18160104

ABSTRACT

The T-cell specific adapter protein (TSAd) encoded by the SH2D2A gene is up-regulated in activated human CD4+ T-cells in a cAMP-dependent manner. Expression of SH2D2A is important for proper activation of T-cells. Here, we show that SH2D2A expression is regulated both at the transcriptional and translational level. cAMP signaling alone induces TSAd-mRNA expression but fails to induce increased TSAd protein levels. By contrast, TCR engagement provides signals for both TSAd transcription and translation. We further show that cAMP signaling can prime T-cells for a more prompt expression of TSAd protein upon TCR stimulation. Our study thus points to a novel mechanism for how cAMP signaling may modulate T-cell activation through transcriptional priming of resting cells.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , CD4-Positive T-Lymphocytes/metabolism , Gene Expression Regulation , Protein Biosynthesis , Transcription, Genetic , CD3 Complex/immunology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/drug effects , Cross-Priming/drug effects , Cross-Priming/immunology , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cytoplasm/drug effects , Cytoplasm/metabolism , Gene Expression Regulation/drug effects , Humans , Isoquinolines/pharmacology , Models, Immunological , Protein Biosynthesis/drug effects , RNA Transport/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Antigen, T-Cell/immunology , Signal Transduction/drug effects , Sulfonamides/pharmacology , Transcription, Genetic/drug effects
8.
Science ; 353(6307): 1549-1552, 2016 09 30.
Article in English | MEDLINE | ID: mdl-27708105

ABSTRACT

Cyclin A2 activates the cyclin-dependent kinases Cdk1 and Cdk2 and is expressed at elevated levels from S phase until early mitosis. We found that mutant mice that cannot elevate cyclin A2 are chromosomally unstable and tumor-prone. Underlying the chromosomal instability is a failure to up-regulate the meiotic recombination 11 (Mre11) nuclease in S phase, which leads to impaired resolution of stalled replication forks, insufficient repair of double-stranded DNA breaks, and improper segregation of sister chromosomes. Unexpectedly, cyclin A2 controlled Mre11 abundance through a C-terminal RNA binding domain that selectively and directly binds Mre11 transcripts to mediate polysome loading and translation. These data reveal cyclin A2 as a mechanistically diverse regulator of DNA replication combining multifaceted kinase-dependent functions with a kinase-independent, RNA binding-dependent role that ensures adequate repair of common replication errors.


Subject(s)
Chromosomal Instability , Cyclin A2/metabolism , DNA Repair Enzymes/genetics , DNA Replication/genetics , DNA-Binding Proteins/genetics , Gene Expression Regulation , Protein Biosynthesis/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Animals , CDC2 Protein Kinase/metabolism , Centrosome/metabolism , Cyclin A2/genetics , DNA Breaks, Double-Stranded , DNA Repair , Humans , Kinesins/metabolism , MRE11 Homologue Protein , Meiosis/genetics , Mice , Mice, Mutant Strains , Mitosis/genetics , RNA, Messenger/genetics , RNA-Binding Proteins/genetics , S Phase/genetics
9.
Immun Inflamm Dis ; 3(2): 103-17, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26029370

ABSTRACT

B lymphopoiesis in bone marrow (BM) is critical for maintaining a diverse peripheral B cell pool to fight infection and establish lifelong immunity. The generation of immature B cells is reduced in Flt3-ligand (FL-/-) mice leading to deficiencies in splenic B cells. Here, we sought to understand the cellular basis of the spleen B cell deficiency in FL-/- mice. Significant reductions in transitional (TS) and follicular (FO) B cells were found in FL-/- mice, and increased frequencies, but not absolute numbers, of marginal zone (MZ) B cells. BAFF-R expression on splenic B cells and serum levels of B cell activating factor (BAFF) was comparable to wildtype (WT) mice. Mixed BM chimeras revealed that the reductions in TS and FO B cells were cell extrinsic. FL administration into FL-/- mice restored the deficiency in TS B cells and normalized the MZ compartment. Ki67 analysis revealed a significant decrease in the proliferative capacity of TS B cells in FL-/- mice. A Bcl2 transgene did not rescue TS cells in FL-/- mice, uncoupling FL-deficiency to Bcl2-dependent survival pathways. Upregulation of CD1d expression and adoptive transfer experiments suggested MZ skewing in FL-/- mice. These findings support an integral role for Flt3 signaling in peripheral B cell maturation.

10.
PLoS One ; 9(1): e85090, 2014.
Article in English | MEDLINE | ID: mdl-24454796

ABSTRACT

Actin depolymerizing factor-homology (ADF-H) family proteins regulate actin filament dynamics at multiple cellular locations. Herein, we have investigated the function of the ADF-H family member coactosin-like 1 (COTL1) in the regulation of actin dynamics at the T cell immune synapse (IS). We initially identified COTL1 in a genetic screen to identify novel regulators of T cell activation, and subsequently found that it associates with F-actin and localizes at the IS in response to TCR+CD28 stimulation. Live cell microscopy showed that depletion of COTL1 protein impaired T cell spreading in response to TCR ligation and abrogated lamellipodial protrusion at the T cell - B cell contact site, producing only a band of F-actin. Significantly, re-expression of wild type COTL1, but not a mutant deficient in F-actin binding could rescue these defects. In addition, COTL1 depletion reduced T cell migration. In vitro studies showed that COTL1 and cofilin compete with each other for binding to F-actin, and COTL1 protects F-actin from cofilin-mediated depolymerization. While depletion of cofilin enhanced F-actin assembly and lamellipodial protrusion at the IS, concurrent depletion of both COTL1 and cofilin restored lamellipodia formation. Taken together, our results suggest that COTL1 regulates lamellipodia dynamics in part by protecting F-actin from cofilin-mediated disassembly.


Subject(s)
Cofilin 1/antagonists & inhibitors , Immunological Synapses/metabolism , Microfilament Proteins/metabolism , Pseudopodia/metabolism , Actins/metabolism , CD28 Antigens/metabolism , Cell Movement/drug effects , Chemokines/pharmacology , Cofilin 1/metabolism , Genetic Complementation Test , Humans , Immunological Synapses/drug effects , Jurkat Cells , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Pseudopodia/drug effects , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
11.
Eur J Immunol ; 35(5): 1612-20, 2005 May.
Article in English | MEDLINE | ID: mdl-15827961

ABSTRACT

T cell-specific adapter protein (TSAd), encoded by the SH2D2A gene, is expressed in activated T cells. The function of TSAd is as yet unknown. We previously showed that TSAd may modulate T cell receptor-triggered signaling events. TSAd contains a Src homology (SH)2 domain, ten tyrosines and a C-terminal proline-rich region. Here, we show that human TSAd interacts with Lck through the Lck SH2 and SH3 domains and is a substrate for Lck. The TSAd C terminus, including the proline-rich region and five tyrosines, is both necessary and sufficient for TSAd interaction with and phosphorylation by Lck. Expression of TSAd in Jurkat TAg cells results in hyperphosphorylation of endogenous Lck on Y394 and to an even larger extent on Y505, resulting in a reduced Y394/Y505 phosphorylation ratio in these cells. Furthermore, full-length TSAd, but not TSAd lacking the C terminus, inhibits the hyperactive Lck Y505F mutant when both are expressed in Jurkat T cells. In contrast, expression of the TSAd C terminus alone is sufficient to inhibit Lck Y505F in phosphorylating its substrates in Jurkat T cells. Our results indicate that the TSAd C terminus is essential for inhibition of Lck activity by TSAd, and suggest a mechanism for how TSAd may inhibit early T cell activation events.


Subject(s)
Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/metabolism , T-Lymphocytes/immunology , Adaptor Proteins, Signal Transducing/immunology , Amino Acid Sequence , Animals , Blotting, Western , Electrophoresis, Polyacrylamide Gel , Humans , Jurkat Cells , Lymphocyte Activation/immunology , Molecular Sequence Data , Phosphorylation , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL